1
|
Jansen I, Cahalane R, Hengst R, Akyildiz A, Farrell E, Gijsen F, Aikawa E, van der Heiden K, Wissing T. The interplay of collagen, macrophages, and microcalcification in atherosclerotic plaque cap rupture mechanics. Basic Res Cardiol 2024; 119:193-213. [PMID: 38329498 PMCID: PMC11008085 DOI: 10.1007/s00395-024-01033-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 02/09/2024]
Abstract
The rupture of an atherosclerotic plaque cap overlying a lipid pool and/or necrotic core can lead to thrombotic cardiovascular events. In essence, the rupture of the plaque cap is a mechanical event, which occurs when the local stress exceeds the local tissue strength. However, due to inter- and intra-cap heterogeneity, the resulting ultimate cap strength varies, causing proper assessment of the plaque at risk of rupture to be lacking. Important players involved in tissue strength include the load-bearing collagenous matrix, macrophages, as major promoters of extracellular matrix degradation, and microcalcifications, deposits that can exacerbate local stress, increasing tissue propensity for rupture. This review summarizes the role of these components individually in tissue mechanics, along with the interplay between them. We argue that to be able to improve risk assessment, a better understanding of the effect of these individual components, as well as their reciprocal relationships on cap mechanics, is required. Finally, we discuss potential future steps, including a holistic multidisciplinary approach, multifactorial 3D in vitro model systems, and advancements in imaging techniques. The obtained knowledge will ultimately serve as input to help diagnose, prevent, and treat atherosclerotic cap rupture.
Collapse
Affiliation(s)
- Imke Jansen
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Rachel Cahalane
- Mechanobiology and Medical Device Research Group (MMDRG), Biomedical Engineering, College of Science and Engineering, University of Galway, Galway, Ireland
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ranmadusha Hengst
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Ali Akyildiz
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Biomechanical Engineering, Technical University Delft, Delft, The Netherlands
| | - Eric Farrell
- Department of Oral and Maxillofacial Surgery, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Frank Gijsen
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Biomechanical Engineering, Technical University Delft, Delft, The Netherlands
| | - Elena Aikawa
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kim van der Heiden
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Tamar Wissing
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
2
|
Abstract
PURPOSE OF REVIEW Aging is an important risk factor for cardiovascular disease and is associated with increased vessel wall stiffness. Pathophysiological stiffening, notably in arteries, disturbs the integrity of the vascular endothelium and promotes permeability and transmigration of immune cells, thereby driving the development of atherosclerosis and related vascular diseases. Effective therapeutic strategies for arterial stiffening are still lacking. RECENT FINDINGS Here, we overview the literature on age-related arterial stiffening, from patient-derived data to preclinical in-vivo and in-vitro findings. First, we overview the common techniques that are used to measure stiffness and discuss the observed stiffness values in atherosclerosis and aging. Next, the endothelial response to stiffening and possibilities to attenuate this response are discussed. SUMMARY Future research that will define the endothelial contribution to stiffness-related cardiovascular disease may provide new targets for intervention to restore endothelial function in atherosclerosis and complement the use of currently applied lipid-lowering, antihypertensive, and anti-inflammatory drugs.
Collapse
Affiliation(s)
- Aukie Hooglugt
- Amsterdam UMC, University of Amsterdam, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences
- Amsterdam UMC, VU University Medical Center, Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Olivia Klatt
- Amsterdam UMC, University of Amsterdam, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences
| | - Stephan Huveneers
- Amsterdam UMC, University of Amsterdam, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences
| |
Collapse
|
3
|
Simon F, Larena-Avellaneda A, Wipper S. Experimental Atherosclerosis Research on Large and Small Animal Models in Vascular Surgery. J Vasc Res 2022; 59:221-228. [PMID: 35760040 PMCID: PMC9533439 DOI: 10.1159/000524795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 04/18/2022] [Indexed: 11/19/2022] Open
Abstract
Animal models have significantly advanced our understanding of the mechanisms of atherosclerosis formation and the evaluation of therapeutic options. The current focus of research is on preventive strategies and includes pharmacologic and biologic interventions directed primarily against smooth-muscle cell proliferation, endovascular devices for recanalization and/or drug delivery, and an integrated approach using both devices and pharmacobiologic agents. The experience over many decades with animal models in vascular research has established that a single, ideal, naturally available model for atherosclerosis does not exist. The spectrum ranges from large animals such as pigs to small animal experiments with genetically modified rodents such as the ApoE-/- mouse with correspondingly differently pronounced changes in their lipid and lipoprotein levels. The development of transgenic variants of currently available models, e.g., an ApoE-deficient rabbit line, has widened our options. Nevertheless, an appreciation of the individual features of natural or stimulated disease in each species is of importance for the proper design and execution of relevant experiments.
Collapse
Affiliation(s)
- Florian Simon
- University Hospital Düsseldorf, Clinic for Vascular and Endovascular Surgery, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Axel Larena-Avellaneda
- Department of Vascular and Endovascular Surgery, Asklepios Clinic Altona, Hamburg, Germany
| | - Sabine Wipper
- Department for Vascular Surgery, University Hospital Innsbruck, Innsbruck, Austria
| |
Collapse
|
4
|
Meneri M, Bonato S, Gagliardi D, Comi GP, Corti S. New Insights into Cerebral Vessel Disease Landscapes at Single-Cell Resolution: Pathogenetic and Therapeutic Perspectives. Biomedicines 2022; 10:1693. [PMID: 35884997 PMCID: PMC9313091 DOI: 10.3390/biomedicines10071693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 07/03/2022] [Accepted: 07/11/2022] [Indexed: 11/19/2022] Open
Abstract
Cerebrovascular diseases are a leading cause of death and disability globally. The development of new therapeutic targets for cerebrovascular diseases (e.g., ischemic, and hemorrhagic stroke, vascular dementia) is limited by a lack of knowledge of the cellular and molecular biology of health and disease conditions and the factors that cause injury to cerebrovascular structures. Here, we describe the role of advances in omics technology, particularly RNA sequencing, in studying high-dimensional, multifaceted profiles of thousands of individual blood and vessel cells at single-cell resolution. This analysis enables the dissection of the heterogeneity of diseased cerebral vessels and their atherosclerotic plaques, including the microenvironment, cell evolutionary trajectory, and immune response pathway. In animal models, RNA sequencing permits the tracking of individual cells (including immunological, endothelial, and vascular smooth muscle cells) that compose atherosclerotic plaques and their alteration under experimental settings such as phenotypic transition. We describe how single-cell RNA transcriptomics in humans allows mapping to the molecular and cellular levels of atherosclerotic plaques in cerebral arteries, tracking individual lymphocytes and macrophages, and how these data can aid in identifying novel immune mechanisms that could be exploited as therapeutic targets for cerebrovascular diseases. Single-cell multi-omics approaches will likely provide the unprecedented resolution and depth of data needed to generate clinically relevant cellular and molecular signatures for the precise treatment of cerebrovascular diseases.
Collapse
Affiliation(s)
- Megi Meneri
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), University of Milan, 20122 Milan, Italy; (M.M.); (D.G.); (G.P.C.)
- Neurology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Sara Bonato
- Stroke Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Delia Gagliardi
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), University of Milan, 20122 Milan, Italy; (M.M.); (D.G.); (G.P.C.)
- Neurology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Giacomo P. Comi
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), University of Milan, 20122 Milan, Italy; (M.M.); (D.G.); (G.P.C.)
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Stefania Corti
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), University of Milan, 20122 Milan, Italy; (M.M.); (D.G.); (G.P.C.)
- Neurology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| |
Collapse
|
5
|
Tissue-engineered collagenous fibrous cap models to systematically elucidate atherosclerotic plaque rupture. Sci Rep 2022; 12:5434. [PMID: 35361847 PMCID: PMC8971478 DOI: 10.1038/s41598-022-08425-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 03/04/2022] [Indexed: 12/24/2022] Open
Abstract
A significant amount of vascular thrombotic events are associated with rupture of the fibrous cap that overlie atherosclerotic plaques. Cap rupture is however difficult to predict due to the heterogenous composition of the plaque, unknown material properties, and the stochastic nature of the event. Here, we aim to create tissue engineered human fibrous cap models with a variable but controllable collagen composition, suitable for mechanical testing, to scrutinize the reciprocal relationships between composition and mechanical properties. Myofibroblasts were cultured in 1 × 1.5 cm-sized fibrin-based constrained gels for 21 days according to established (dynamic) culture protocols (i.e. static, intermittent or continuous loading) to vary collagen composition (e.g. amount, type and organization). At day 7, a soft 2 mm ∅ fibrin inclusion was introduced in the centre of each tissue to mimic the soft lipid core, simulating the heterogeneity of a plaque. Results demonstrate reproducible collagenous tissues, that mimic the bulk mechanical properties of human caps and vary in collagen composition due to the presence of a successfully integrated soft inclusion and the culture protocol applied. The models can be deployed to assess tissue mechanics, evolution and failure of fibrous caps or complex heterogeneous tissues in general.
Collapse
|
6
|
Hill CA, Fernandez DM, Giannarelli C. Single cell analyses to understand the immune continuum in atherosclerosis. Atherosclerosis 2021; 330:85-94. [PMID: 33934886 PMCID: PMC8855459 DOI: 10.1016/j.atherosclerosis.2021.04.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 03/22/2021] [Accepted: 04/08/2021] [Indexed: 12/13/2022]
Abstract
Atherosclerosis is initiated by the accumulation of lipids in the arterial wall that trigger a complex and poorly understood network of inflammatory processes. At the same time, recent clinical findings reveal that targeting specific immune alterations in patients with cardiovascular disease (CVD) represents a promising approach to preventing recurrent cardiovascular events. In order to achieve these tailored therapies, it is critical to resolve the heterogenous environment of the atherosclerotic lesion and decipher the complex structural and functional changes which immune cells undergo throughout disease progression. Recently, single-cell approaches including single cell mass cytometry by time of flight (CyTOF), single cell RNA sequencing (scRNA-seq) and Cellular Indexing of Transcriptomes and Epitopes by Sequencing (CITE-seq) have emerged as valuable tools in resolving cellular plasticity within atherosclerotic lesions. In this review, we will discuss the most important insights that have been gleaned from the application of these single-cell approaches to validated experimental models of atherosclerosis. Additionally, as clinical progress in treatment of the disease depends on the translation of discoveries to human tissues, we will also examine the challenges associated with the application of single-cell approaches to human vascular tissue and the discoveries made by the initial efforts in this direction. Finally, we will analyze the advantages and limitations of dissociative single-cell approaches and how novel in-situ technologies could advance the field by allowing for the investigation of individual cells while preserving the heterogenous architecture of the atherosclerotic lesion.
Collapse
Affiliation(s)
| | | | - Chiara Giannarelli
- Department of Medicine, Cardiovascular Research Center, USA; Department of Genetics and Genomic Sciences, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medicine, Division of Cardiology and Cardiovascular Research Center at NYU Langone NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
7
|
Hoogendoorn A, Avery TD, Li J, Bursill C, Abell A, Grace PM. Emerging Therapeutic Applications for Fumarates. Trends Pharmacol Sci 2021; 42:239-254. [PMID: 33618840 PMCID: PMC7954891 DOI: 10.1016/j.tips.2021.01.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 01/14/2021] [Accepted: 01/20/2021] [Indexed: 02/08/2023]
Abstract
Fumarates are successfully used for the treatment of psoriasis and multiple sclerosis. Their antioxidative, immunomodulatory, and neuroprotective properties make fumarates attractive therapeutic candidates for other pathologies. The exact working mechanisms of fumarates are, however, not fully understood. Further elucidation of the mechanisms is required if these drugs are to be successfully repurposed for other diseases. Towards this, administration route, dosage, and treatment timing, frequency, and duration are important parameters to consider and optimize with clinical paradigms in mind. Here, we summarize the rapidly expanding literature on the pharmacokinetics and pharmacodynamics of fumarates, including a discussion on two recently FDA-approved fumarates VumerityTM and BafiertamTM. We review emerging applications of fumarates, focusing on neurological and cardiovascular diseases.
Collapse
Affiliation(s)
- Ayla Hoogendoorn
- Vascular and Heart Health, Life Long Health Theme, South Australian Health and Medical Research Institute, Adelaide, Australia; ARC Centre of Excellence for Nanoscale BioPhotonics, The University of Adelaide, Australia.
| | - Thomas D Avery
- ARC Centre of Excellence for Nanoscale BioPhotonics, The University of Adelaide, Australia; Institute for Photonics and Advanced Sensing & Department of Chemistry, The University of Adelaide, Australia
| | - Jiahe Li
- Laboratories of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christina Bursill
- Vascular and Heart Health, Life Long Health Theme, South Australian Health and Medical Research Institute, Adelaide, Australia; ARC Centre of Excellence for Nanoscale BioPhotonics, The University of Adelaide, Australia; Faculty of Health and Medical Science, University of Adelaide, Adelaide, Australia
| | - Andrew Abell
- ARC Centre of Excellence for Nanoscale BioPhotonics, The University of Adelaide, Australia; Institute for Photonics and Advanced Sensing & Department of Chemistry, The University of Adelaide, Australia
| | - Peter M Grace
- Laboratories of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
8
|
Atherosclerosis in Different Vascular Locations Unbiasedly Approached with Mouse Genetics. Genes (Basel) 2020; 11:genes11121427. [PMID: 33260687 PMCID: PMC7760563 DOI: 10.3390/genes11121427] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/24/2020] [Accepted: 11/26/2020] [Indexed: 01/01/2023] Open
Abstract
Atherosclerosis in different vascular locations leads to distinct clinical consequences, such as ischemic stroke and myocardial infarction. Genome-wide association studies in humans revealed that genetic loci responsible for carotid plaque and coronary artery disease were not overlapping, suggesting that distinct genetic pathways might be involved for each location. While elevated plasma cholesterol is a common risk factor, plaque development in different vascular beds is influenced by hemodynamics and intrinsic vascular integrity. Despite the limitation of species differences, mouse models provide platforms for unbiased genetic approaches. Mouse strain differences also indicate that susceptibility to atherosclerosis varies, depending on vascular locations, and that the location specificity is genetically controlled. Quantitative trait loci analyses in mice suggested candidate genes, including Mertk and Stab2, although how each gene affects the location-specific atherosclerosis needs further elucidation. Another unbiased approach of single-cell transcriptome analyses revealed the presence of a small subpopulation of vascular smooth muscle cells (VSMCs), which are “hyper-responsive” to inflammatory stimuli. These cells are likely the previously-reported Sca1+ progenitor cells, which can differentiate into multiple lineages in plaques. Further spatiotemporal analyses of the progenitor cells are necessary, since their distribution pattern might be associated with the location-dependent plaque development.
Collapse
|
9
|
Fernandez DM, Rahman AH, Fernandez NF, Chudnovskiy A, Amir EAD, Amadori L, Khan NS, Wong CK, Shamailova R, Hill CA, Wang Z, Remark R, Li JR, Pina C, Faries C, Awad AJ, Moss N, Bjorkegren JLM, Kim-Schulze S, Gnjatic S, Ma'ayan A, Mocco J, Faries P, Merad M, Giannarelli C. Single-cell immune landscape of human atherosclerotic plaques. Nat Med 2019; 25:1576-1588. [PMID: 31591603 PMCID: PMC7318784 DOI: 10.1038/s41591-019-0590-4] [Citation(s) in RCA: 548] [Impact Index Per Article: 109.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 08/16/2019] [Indexed: 12/29/2022]
Abstract
Atherosclerosis is driven by multifaceted contributions of the immune system within the circulation and at vascular focal sites. However, specific characteristics of dysregulated immune cells within atherosclerotic lesions that lead to clinical events such as ischemic stroke or myocardial infarction are poorly understood. Here, using single-cell proteomic and transcriptomic analyses, we uncovered distinct features of both T cells and macrophages in carotid artery plaques of patients with clinically symptomatic disease (recent stroke or transient ischemic attack) compared to asymptomatic disease (no recent stroke). Plaques from symptomatic patients were characterized by a distinct subset of CD4+ T cells and by T cells that were activated and differentiated. Moreover, some T cell subsets in these plaques presented markers of T cell exhaustion. Additionally, macrophages from these plaques contained alternatively activated phenotypes, including subsets associated with plaque vulnerability. In plaques from asymptomatic patients, T cells and macrophages were activated and displayed evidence of interleukin-1β signaling. The identification of specific features of innate and adaptive immune cells in plaques that are associated with cerebrovascular events may enable the design of more precisely tailored cardiovascular immunotherapies.
Collapse
Affiliation(s)
- Dawn M Fernandez
- Cardiovascular Research Center, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adeeb H Rahman
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nicolas F Fernandez
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aleksey Chudnovskiy
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - El-Ad David Amir
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Letizia Amadori
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nayaab S Khan
- Cardiovascular Research Center, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christine K Wong
- Cardiovascular Research Center, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Roza Shamailova
- Cardiovascular Research Center, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christopher A Hill
- Cardiovascular Research Center, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zichen Wang
- Mount Sinai Center for Bioinformatics, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Romain Remark
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Innate Pharma, Marseille, France
| | - Jennifer R Li
- Department of Surgery, Vascular Division, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christian Pina
- Department of Surgery, Vascular Division, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christopher Faries
- Department of Surgery, Vascular Division, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ahmed J Awad
- Cerebrovascular Center, Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Noah Moss
- Cardiovascular Research Center, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Johan L M Bjorkegren
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Integrated Cardio MetabolicCentre, Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset, Huddinge, Sweden
| | - Seunghee Kim-Schulze
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Hematology and Medical Oncology Division, The Tish Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sacha Gnjatic
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Avi Ma'ayan
- Mount Sinai Center for Bioinformatics, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - J Mocco
- Cerebrovascular Center, Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Peter Faries
- Department of Surgery, Vascular Division, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Merad
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chiara Giannarelli
- Cardiovascular Research Center, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
10
|
Meester EJ, Krenning BJ, de Swart J, Segbers M, Barrett HE, Bernsen MR, Van der Heiden K, de Jong M. Perspectives on Small Animal Radionuclide Imaging; Considerations and Advances in Atherosclerosis. Front Med (Lausanne) 2019; 6:39. [PMID: 30915335 PMCID: PMC6421263 DOI: 10.3389/fmed.2019.00039] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 02/11/2019] [Indexed: 12/20/2022] Open
Abstract
This review addresses nuclear SPECT and PET imaging in small animals in relation to the atherosclerotic disease process, one of our research topics of interest. Imaging of atherosclerosis in small animal models is challenging, as it operates at the limits of current imaging possibilities regarding sensitivity, and spatial resolution. Several topics are discussed, including technical considerations that apply to image acquisition, reconstruction, and analysis. Moreover, molecules developed for or applied in these small animal nuclear imaging studies are listed, including target-directed molecules, useful for imaging organs or tissues that have elevated expression of the target compared to other tissues, and molecules that serve as substrates for metabolic processes. Differences between animal models and human pathophysiology that should be taken into account during translation from animal to patient as well as differences in tracer behavior in animal vs. man are also described. Finally, we give a future outlook on small animal radionuclide imaging in atherosclerosis, followed by recommendations. The challenges and solutions described might be applicable to other research fields of health and disease as well.
Collapse
Affiliation(s)
- Eric J Meester
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, Netherlands.,Department of Biomedical Engineering, Thorax Center, Erasmus Medical Center, Rotterdam, Netherlands
| | - B J Krenning
- Department of Cardiology, Thorax Center, Erasmus Medical Center, Rotterdam, Netherlands
| | - J de Swart
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, Netherlands
| | - M Segbers
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, Netherlands
| | - H E Barrett
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, Netherlands.,Department of Biomedical Engineering, Thorax Center, Erasmus Medical Center, Rotterdam, Netherlands
| | - M R Bernsen
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, Netherlands
| | - K Van der Heiden
- Department of Biomedical Engineering, Thorax Center, Erasmus Medical Center, Rotterdam, Netherlands
| | - Marion de Jong
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, Netherlands
| |
Collapse
|
11
|
Millar CL, Norris GH, Jiang C, Kry J, Vitols A, Garcia C, Park YK, Lee JY, Blesso CN. Long-Term Supplementation of Black Elderberries Promotes Hyperlipidemia, but Reduces Liver Inflammation and Improves HDL Function and Atherosclerotic Plaque Stability in Apolipoprotein E-Knockout Mice. Mol Nutr Food Res 2018; 62:e1800404. [PMID: 30267603 DOI: 10.1002/mnfr.201800404] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 09/19/2018] [Indexed: 11/08/2022]
Abstract
SCOPE HDL particles are protective against atherosclerosis, but may become dysfunctional during inflammation and chronic disease progression. Anthocyanin-rich foods, such as the black elderberry, may improve HDL function and prevent disease development via antioxidant and/or anti-inflammatory effects. This study investigates the long-term consumption of black elderberry extract (BEE) on HDL function and atherosclerosis in apolipoprotein (apo) E-/- mice. METHODS AND RESULTS ApoE-/- mice (n = 12/group) are fed a low-fat diet, supplemented with 0, 0.25%, or 1% (by weight) BEE (≈37.5-150 mg anthocyanins per kg body weight) for 24 weeks. Feeding 1% BEE increases total serum cholesterol (+31%) and non-HDL cholesterol (+32%) compared with the control diet. PON1 arylesterase (+32%) and lactonase (+45%) activities also increase with the 1% BEE diet. Both 0.25% BEE and 1% BEE diets strongly increase HDL cholesterol efflux capacity (CEC) by 64% and 85%, respectively. Further, BEE dose-dependently lowers serum liver enzymes and hepatic inflammatory gene expression. Although there is no change in neutral lipid accumulation in atherosclerotic lesions, BEE promotes connective tissue deposition in the aortic root. CONCLUSIONS Chronic BEE supplementation in apoE-/- mice dose-dependently improves HDL function. Despite BEE promoting hyperlipidemia, which likely offsets HDL effects, BEE increases connective tissue content, suggesting improved atherosclerotic plaque stability.
Collapse
Affiliation(s)
- Courtney L Millar
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT, 06118, USA
| | - Gregory H Norris
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT, 06118, USA
| | - Christina Jiang
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT, 06118, USA
| | - James Kry
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT, 06118, USA
| | - Addison Vitols
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT, 06118, USA
| | - Chelsea Garcia
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT, 06118, USA
| | - Young-Ki Park
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT, 06118, USA
| | - Ji-Young Lee
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT, 06118, USA
| | - Christopher N Blesso
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT, 06118, USA
| |
Collapse
|
12
|
Caporali A, Bäck M, Daemen MJ, Hoefer IE, Jones EA, Lutgens E, Matter CM, Bochaton-Piallat ML, Siekmann AF, Sluimer JC, Steffens S, Tuñón J, Vindis C, Wentzel JJ, Ylä-Herttuala S, Evans PC. Future directions for therapeutic strategies in post-ischaemic vascularization: a position paper from European Society of Cardiology Working Group on Atherosclerosis and Vascular Biology. Cardiovasc Res 2018; 114:1411-1421. [PMID: 30016405 PMCID: PMC6106103 DOI: 10.1093/cvr/cvy184] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 05/16/2018] [Accepted: 07/16/2018] [Indexed: 12/16/2022] Open
Abstract
Modulation of vessel growth holds great promise for treatment of cardiovascular disease. Strategies to promote vascularization can potentially restore function in ischaemic tissues. On the other hand, plaque neovascularization has been shown to associate with vulnerable plaque phenotypes and adverse events. The current lack of clinical success in regulating vascularization illustrates the complexity of the vascularization process, which involves a delicate balance between pro- and anti-angiogenic regulators and effectors. This is compounded by limitations in the models used to study vascularization that do not reflect the eventual clinical target population. Nevertheless, there is a large body of evidence that validate the importance of angiogenesis as a therapeutic concept. The overall aim of this Position Paper of the ESC Working Group of Atherosclerosis and Vascular biology is to provide guidance for the next steps to be taken from pre-clinical studies on vascularization towards clinical application. To this end, the current state of knowledge in terms of therapeutic strategies for targeting vascularization in post-ischaemic disease is reviewed and discussed. A consensus statement is provided on how to optimize vascularization studies for the identification of suitable targets, the use of animal models of disease, and the analysis of novel delivery methods.
Collapse
Affiliation(s)
- Andrea Caporali
- University/British Heart Foundation Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Magnus Bäck
- Division of Valvular and Coronary Disease, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet and University Hospital Stockholm, Stockholm, Sweden
- INSERM U1116, University of Lorraine, Nancy University Hospital, Nancy, France
| | - Mat J Daemen
- Department of Pathology, Academic Medical Hospital, University of Amsterdam, Amsterdam, The Netherlands
| | - Imo E Hoefer
- Laboratory of Experimental Cardiology and Laboratory of Clinical Chemistry and Hematology, UMC Utrecht, Utrecht, Netherlands
| | | | - Esther Lutgens
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Christian M Matter
- Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland
| | | | - Arndt F Siekmann
- Max Planck Institute for Molecular Biomedicine, Muenster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003–CiM), University of Muenster, Muenster, Germany
| | - Judith C Sluimer
- University/British Heart Foundation Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Department of Pathology, CARIM, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Sabine Steffens
- Ludwig-Maximilians-University, German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - José Tuñón
- IIS-Fundación Jiménez Díaz, Madrid, Spain
- Autónoma University, Madrid, Spain
| | - Cecile Vindis
- INSERM U1048/Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
| | - Jolanda J Wentzel
- Department of Cardiology, Biomechanics Laboratory, Erasmus MC, Rotterdam, The Netherlands
| | - Seppo Ylä-Herttuala
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
- Heart Center and Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland
| | - Paul C Evans
- Department of Infection, Immunity and Cardiovascular Disease, Faculty of Medicine, Dentistry and Health, the INSIGNEO Institute for In Silico Medicine and the Bateson Centre, University of Sheffield, Sheffield, UK
| |
Collapse
|
13
|
Xing R, Moerman AM, Ridwan Y, Daemen MJ, van der Steen AFW, Gijsen FJH, van der Heiden K. Temporal and spatial changes in wall shear stress during atherosclerotic plaque progression in mice. ROYAL SOCIETY OPEN SCIENCE 2018; 5:171447. [PMID: 29657758 PMCID: PMC5882682 DOI: 10.1098/rsos.171447] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 02/07/2018] [Indexed: 05/03/2023]
Abstract
Wall shear stress (WSS) is involved in atherosclerotic plaque initiation, yet its role in plaque progression remains unclear. We aimed to study (i) the temporal and spatial changes in WSS over a growing plaque and (ii) the correlation between WSS and plaque composition, using animal-specific data in an atherosclerotic mouse model. Tapered casts were placed around the right common carotid arteries (RCCA) of ApoE-/- mice. At 5, 7 and 9 weeks after cast placement, RCCA geometry was reconstructed using contrast-enhanced micro-CT. Lumen narrowing was observed in all mice, indicating the progression of a lumen intruding plaque. Next, we determined the flow rate in the RCCA of each mouse using Doppler Ultrasound and computed WSS at all time points. Over time, as the plaque developed and further intruded into the lumen, absolute WSS significantly decreased. Finally at week 9, plaque composition was histologically characterized. The proximal part of the plaque was small and eccentric, exposed to relatively lower WSS. Close to the cast a larger and concentric plaque was present, exposed to relatively higher WSS. Lower WSS was significantly correlated to the accumulation of macrophages in the eccentric plaque. When pooling data of all animals, correlation between WSS and plaque composition was weak and no longer statistically significant. In conclusion, our data showed that in our mouse model absolute WSS strikingly decreased during disease progression, which was significantly correlated to plaque area and macrophage content. Besides, our study demonstrates the necessity to analyse individual animals and plaques when studying correlations between WSS and plaque composition.
Collapse
Affiliation(s)
- R. Xing
- Department of Biomedical Engineering, Thorax Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - A. M. Moerman
- Department of Biomedical Engineering, Thorax Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Y. Ridwan
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - M. J. Daemen
- Department of Pathology, Academic Medical Center, Amsterdam, The Netherlands
| | - A. F. W. van der Steen
- Department of Biomedical Engineering, Thorax Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - F. J. H. Gijsen
- Department of Biomedical Engineering, Thorax Center, Erasmus University Medical Center, Rotterdam, The Netherlands
- Authors for correspondence: F. J. H. Gijsen e-mail:
| | - K. van der Heiden
- Department of Biomedical Engineering, Thorax Center, Erasmus University Medical Center, Rotterdam, The Netherlands
- Authors for correspondence: K. van der Heiden e-mail:
| |
Collapse
|
14
|
Robinet P, Milewicz DM, Cassis LA, Leeper NJ, Lu HS, Smith JD. Consideration of Sex Differences in Design and Reporting of Experimental Arterial Pathology Studies-Statement From ATVB Council. Arterioscler Thromb Vasc Biol 2018; 38:292-303. [PMID: 29301789 DOI: 10.1161/atvbaha.117.309524] [Citation(s) in RCA: 197] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 12/20/2017] [Indexed: 12/15/2022]
Abstract
There are many differences in arterial diseases between men and women, including prevalence, clinical manifestations, treatments, and prognosis. The new policy of the National Institutes of Health, which requires the inclusion of sex as a biological variable for preclinical studies, aims to foster new mechanistic insights and to enhance our understanding of sex differences in human diseases. The purpose of this statement is to suggest guidelines for designing and reporting sex as a biological variable in animal models of atherosclerosis, thoracic and abdominal aortic aneurysms, and peripheral arterial disease. We briefly review sex differences of these human diseases and their animal models, followed by suggestions on experimental design and reporting of animal studies for these vascular pathologies.
Collapse
Affiliation(s)
- Peggy Robinet
- From the Department of Cellular and Molecular Medicine, Cleveland Clinic, OH (P.R., J.D.S.); Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston (D.M.M.); Department of Pharmacology and Nutritional Sciences (L.A.C.) and Saha Cardiovascular Research Center and Department of Physiology (H.S.L.), University of Kentucky, Lexington; and Division of Vascular Surgery, Department of Surgery, Stanford University, CA (N.J.L.)
| | - Dianna M Milewicz
- From the Department of Cellular and Molecular Medicine, Cleveland Clinic, OH (P.R., J.D.S.); Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston (D.M.M.); Department of Pharmacology and Nutritional Sciences (L.A.C.) and Saha Cardiovascular Research Center and Department of Physiology (H.S.L.), University of Kentucky, Lexington; and Division of Vascular Surgery, Department of Surgery, Stanford University, CA (N.J.L.)
| | - Lisa A Cassis
- From the Department of Cellular and Molecular Medicine, Cleveland Clinic, OH (P.R., J.D.S.); Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston (D.M.M.); Department of Pharmacology and Nutritional Sciences (L.A.C.) and Saha Cardiovascular Research Center and Department of Physiology (H.S.L.), University of Kentucky, Lexington; and Division of Vascular Surgery, Department of Surgery, Stanford University, CA (N.J.L.)
| | - Nicholas J Leeper
- From the Department of Cellular and Molecular Medicine, Cleveland Clinic, OH (P.R., J.D.S.); Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston (D.M.M.); Department of Pharmacology and Nutritional Sciences (L.A.C.) and Saha Cardiovascular Research Center and Department of Physiology (H.S.L.), University of Kentucky, Lexington; and Division of Vascular Surgery, Department of Surgery, Stanford University, CA (N.J.L.)
| | - Hong S Lu
- From the Department of Cellular and Molecular Medicine, Cleveland Clinic, OH (P.R., J.D.S.); Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston (D.M.M.); Department of Pharmacology and Nutritional Sciences (L.A.C.) and Saha Cardiovascular Research Center and Department of Physiology (H.S.L.), University of Kentucky, Lexington; and Division of Vascular Surgery, Department of Surgery, Stanford University, CA (N.J.L.)
| | - Jonathan D Smith
- From the Department of Cellular and Molecular Medicine, Cleveland Clinic, OH (P.R., J.D.S.); Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston (D.M.M.); Department of Pharmacology and Nutritional Sciences (L.A.C.) and Saha Cardiovascular Research Center and Department of Physiology (H.S.L.), University of Kentucky, Lexington; and Division of Vascular Surgery, Department of Surgery, Stanford University, CA (N.J.L.).
| |
Collapse
|
15
|
Lip GYH, Weber C. Editors’ Choice 2016 papers in Thrombosis and Haemostasis. Thromb Haemost 2017; 117:204-206. [DOI: 10.1160/th16-11-0899] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 11/30/2016] [Indexed: 11/05/2022]
|
16
|
CD80 Is Upregulated in a Mouse Model with Shear Stress-Induced Atherosclerosis and Allows for Evaluating CD80-Targeting PET Tracers. Mol Imaging Biol 2017; 19:90-99. [PMID: 27430577 DOI: 10.1007/s11307-016-0987-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
PURPOSE A shear stress-induced atherosclerosis mouse model was characterized for its expression of inflammation markers with focus on CD80. With this model, we evaluated two positron emission tomography (PET) radiotracers targeting CD80 as well as 2-deoxy-2-[18F]fluoro-D-mannose ([18F]FDM) in comparison with 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG). PROCEDURE A flow constrictive cuff implanted around the common carotid artery in apolipoprotein E knockout mice resulted in plaque formation. CD80 expression levels and plaque histopathology were evaluated. Serial PET/X-ray computed tomography scans were performed to follow inflammation. RESULTS Plaque formation with increased levels of CD80 was observed. Histologically, plaques presented macrophage-rich and large necrotic areas covered by a thin fibrous cap. Of the CD80-specific tracers, one displayed an increased uptake in plaques by PET. Both [18F]FDG and [18F]FDM accumulated in atherosclerotic plaques. CONCLUSION This mouse model presented, similar to humans, an increased expression of CD80 which renders it suitable for non-invasively targeting CD80-positive immune cells and evaluating CD80-specific radiotracers.
Collapse
|
17
|
de Vries MR, Quax PHA. Plaque angiogenesis and its relation to inflammation and atherosclerotic plaque destabilization. Curr Opin Lipidol 2016; 27:499-506. [PMID: 27472406 DOI: 10.1097/mol.0000000000000339] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW The review discusses the recent literature on plaque angiogenesis and its relation to inflammation and plaque destabilization. Furthermore, it discusses how plaque angiogenesis can be used to monitor atherosclerosis and serve as a therapeutic target. RECENT FINDINGS Histopathologic studies have shown a clear relationship between plaque angiogenesis, intraplaque hemorrhage (IPH), plaque vulnerability, and cardiovascular events. Hypoxia is a main driver of plaque angiogenesis and the mechanism behind angiogenesis is only partly known. IPH, as the result of immature neovessels, is associated with increased influx of inflammatory cells in the plaques. Experimental models displaying certain features of human atherosclerosis such as plaque angiogenesis or IPH are developed and can contribute to unraveling the mechanism behind plaque vulnerability. New imaging techniques are established, with which plaque angiogenesis and vulnerability can be detected. Furthermore, antiangiogenic therapies in atherosclerosis gain much attention. SUMMARY Plaque angiogenesis, IPH, and inflammation contribute to plaque vulnerability. Histopathologic and imaging studies together with specific experimental studies have provided insights in plaque angiogenesis and plaque vulnerability. However, more extensive knowledge on the underlying mechanism is required for establishing new therapies for patients at risk.
Collapse
Affiliation(s)
- Margreet R de Vries
- Department of Surgery, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
18
|
Molecular Imaging of Vulnerable Atherosclerotic Plaques in Animal Models. Int J Mol Sci 2016; 17:ijms17091511. [PMID: 27618031 PMCID: PMC5037788 DOI: 10.3390/ijms17091511] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 08/24/2016] [Accepted: 08/31/2016] [Indexed: 12/13/2022] Open
Abstract
Atherosclerosis is characterized by intimal plaques of the arterial vessels that develop slowly and, in some cases, may undergo spontaneous rupture with subsequent heart attack or stroke. Currently, noninvasive diagnostic tools are inadequate to screen atherosclerotic lesions at high risk of acute complications. Therefore, the attention of the scientific community has been focused on the use of molecular imaging for identifying vulnerable plaques. Genetically engineered murine models such as ApoE−/− and ApoE−/−Fbn1C1039G+/− mice have been shown to be useful for testing new probes targeting biomarkers of relevant molecular processes for the characterization of vulnerable plaques, such as vascular endothelial growth factor receptor (VEGFR)-1, VEGFR-2, intercellular adhesion molecule (ICAM)-1, P-selectin, and integrins, and for the potential development of translational tools to identify high-risk patients who could benefit from early therapeutic interventions. This review summarizes the main animal models of vulnerable plaques, with an emphasis on genetically altered mice, and the state-of-the-art preclinical molecular imaging strategies.
Collapse
|
19
|
Evans PC, Gijsen FJH, Wentzel JJ, van der Heiden K. Biomechanics in vascular biology and cardiovascular disease. Thromb Haemost 2016; 115:465-6. [PMID: 26864973 DOI: 10.1160/th16-01-0075] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 02/01/2016] [Indexed: 11/05/2022]
Affiliation(s)
- Paul C Evans
- Paul Evans, Sheffield University, Sheffield S10 2RX, UK, Tel.: +44 1142712052, Fax: +44 1142712052, E-mail:
| | | | | | | |
Collapse
|